Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Eur J Pharm Sci ; 184: 106418, 2023 May 01.
Artículo en Inglés | MEDLINE | ID: covidwho-2291727

RESUMEN

INTRODUCTION: In the randomized double-blind placebo-controlled CounterCOVID study, oral imatinib treatment conferred a positive clinical outcome and a signal for reduced mortality in COVID-19 patients. High concentrations of alpha-1 acid glycoprotein (AAG) were observed in these patients and were associated with increased total imatinib concentrations. AIMS: This post-hoc study aimed to compare the difference in exposure following oral imatinib administration in COVID-19 patients to cancer patients and assess assocations between pharmacokinetic (PK) parameters and pharmacodynamic (PD) outcomes of imatinib in COVID-19 patients. We hypothesize that a relatively higher drug exposure of imatinib in severe COVID-19 patients leads to improved pharmacodynamic outcome parameters. METHODS: 648 total concentration plasma samples obtained from 168 COVID-19 patients were compared to 475 samples of 105 cancer patients, using an AAG-binding model. Total trough concentration at steady state (Cttrough) and total average area under the concentration-time curve (AUCtave) were associated with ratio between partial oxygen pressure and fraction of inspired oxygen (P/F), WHO ordinal scale (WHO-score) and liberation of oxygen supplementation (O2lib). Linear regression, linear mixed effects models and time-to-event analysis were adjusted for possible confounders. RESULTS: AUCtave and Cttrough were respectively 2.21-fold (95%CI 2.07-2.37) and 1.53-fold (95%CI 1.44-1.63) lower for cancer compared to COVID-19 patients. Cttrough, not AUCtave, associated significantly with P/F (ß=-19,64; p-value=0.014) and O2lib (HR 0.78; p-value= 0.032), after adjusting for sex, age, neutrophil-lymphocyte ratio, dexamethasone concomitant treatment, AAG and baseline P/F-and WHO-score. Cttrough, but not AUCtave associated significantly with WHO-score. These results suggest an inverse relationship between PK-parameters, Cttrough and AUCtave, and PD outcomes. CONCLUSION: COVID-19 patients exhibit higher total imatinib exposure compared to cancer patients, attributed to differences in plasma protein concentrations. Higher imatinib exposure in COVID-19 patients did not associate with improved clinical outcomes. Cttrough and AUCtave inversely associated with some PD-outcomes, which may be biased by disease course, variability in metabolic rate and protein binding. Therefore, additional PKPD analyses into unbound imatinib and its main metabolite may better explain exposure-response.


Asunto(s)
COVID-19 , Neoplasias , Humanos , Mesilato de Imatinib/farmacología , Mesilato de Imatinib/uso terapéutico , Reposicionamiento de Medicamentos , Neoplasias/tratamiento farmacológico
2.
Cell Mol Immunol ; 20(4): 351-364, 2023 04.
Artículo en Inglés | MEDLINE | ID: covidwho-2287148

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced cytokine storm is closely associated with coronavirus disease 2019 (COVID-19) severity and lethality. However, drugs that are effective against inflammation to treat lethal COVID-19 are still urgently needed. Here, we constructed a SARS-CoV-2 spike protein-specific CAR, and human T cells infected with this CAR (SARS-CoV-2-S CAR-T) and stimulated with spike protein mimicked the T-cell responses seen in COVID-19 patients, causing cytokine storm and displaying a distinct memory, exhausted, and regulatory T-cell phenotype. THP1 remarkably augmented cytokine release in SARS-CoV-2-S CAR-T cells when they were in coculture. Based on this "two-cell" (CAR-T and THP1 cells) model, we screened an FDA-approved drug library and found that felodipine, fasudil, imatinib, and caspofungin were effective in suppressing the release of cytokines, which was likely due to their ability to suppress the NF-κB pathway in vitro. Felodipine, fasudil, imatinib, and caspofungin were further demonstrated, although to different extents, to attenuate lethal inflammation, ameliorate severe pneumonia, and prevent mortality in a SARS-CoV-2-infected Syrian hamster model, which were also linked to their suppressive role in inflammation. In summary, we established a SARS-CoV-2-specific CAR-T-cell model that can be utilized as a tool for anti-inflammatory drug screening in a fast and high-throughput manner. The drugs identified herein have great potential for early treatment to prevent COVID-19 patients from cytokine storm-induced lethality in the clinic because they are safe, inexpensive, and easily accessible for immediate use in most countries.


Asunto(s)
COVID-19 , Receptores Quiméricos de Antígenos , Humanos , SARS-CoV-2/metabolismo , Mesilato de Imatinib/farmacología , Mesilato de Imatinib/uso terapéutico , Caspofungina , Felodipino , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Inflamación , Citocinas/metabolismo
3.
Microbiol Spectr ; 11(3): e0510522, 2023 Jun 15.
Artículo en Inglés | MEDLINE | ID: covidwho-2263704

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19). SARS-CoV-2 propagation is mediated by the protein interaction between viral proteins and host cells. Tyrosine kinase has been implicated in viral replication, and hence, it has become a target for developing antiviral drugs. We have previously reported that receptor tyrosine kinase inhibitor blocks the replication of hepatitis C virus (HCV). In the present study, we investigated two receptor tyrosine kinase-specific inhibitors, amuvatinib and imatinib, for their potential antiviral efficacies against SARS-CoV-2. Treatment with either amuvatinib or imatinib displays an effective inhibitory activity against SARS-CoV-2 propagation without an obvious cytopathic effect in Vero E6 cells. Notably, amuvatinib exerts a stronger antiviral activity than imatinib against SARS-CoV-2 infection. Amuvatinib blocks SARS-CoV-2 infection with a 50% effective concentration (EC50) value ranging from ~0.36 to 0.45 µM in Vero E6 cells. We further demonstrate that amuvatinib inhibits SARS-CoV-2 propagation in human lung Calu-3 cells. Using pseudoparticle infection assay, we verify that amuvatinib blocks SARS-CoV-2 at the entry step of the viral life cycle. More specifically, amuvatinib inhibits SARS-CoV-2 infection at the binding-attachment step. Moreover, amuvatinib exhibits highly efficient antiviral activity against emerging SARS-CoV-2 variants. Importantly, we demonstrate that amuvatinib inhibits SARS-CoV-2 infection by blocking ACE2 cleavage. Taken together, our data suggest that amuvatinib may provide a potential therapeutic agent for the treatment of COVID-19. IMPORTANCE Tyrosine kinase has been implicated in viral replication and has become an antiviral drug target. Here, we chose two well-known receptor tyrosine kinase inhibitors, amuvatinib and imatinib, and evaluated their drug potencies against SARS-CoV-2. Surprisingly, amuvatinib displays a stronger antiviral activity than imatinib against SARS-CoV-2. Amuvatinib blocks SARS-CoV-2 infection by inhibiting ACE2 cleavage and the subsequent soluble ACE2 receptor. All these data suggest that amuvatinib may be a potential therapeutic agent in SARS-CoV-2 prevention for those experiencing vaccine breakthroughs.


Asunto(s)
COVID-19 , Animales , Humanos , SARS-CoV-2 , Mesilato de Imatinib/farmacología , Mesilato de Imatinib/uso terapéutico , Enzima Convertidora de Angiotensina 2 , Antivirales/farmacología , Antivirales/uso terapéutico , Proteínas Tirosina Quinasas/farmacología , Estadios del Ciclo de Vida
4.
Int J Mol Sci ; 24(3)2023 Feb 02.
Artículo en Inglés | MEDLINE | ID: covidwho-2225333

RESUMEN

There are a number of uncertainties regarding plasma protein binding and blood distribution of the active drugs favipiravir (FAVI), molnupiravir (MOLNU) and imatinib (IMA), which were recently proposed as therapeutics for the treatment of COVID-19 disease. Therefore, proton dissociation processes, solubility, lipophilicity, and serum protein binding of these three substances were investigated in detail. The drugs display various degrees of lipophilicity at gastric (pH 2.0) and blood pH (pH 7.4). The determined pKa values explain well the changes in lipophilic character of the respective compounds. The serum protein binding was studied by membrane ultrafiltration, frontal analysis capillary electrophoresis, steady-state fluorometry, and fluorescence anisotropy techniques. The studies revealed that the ester bond in MOLNU is hydrolyzed by protein constituents of blood serum. Molnupiravir and its hydrolyzed form do not bind considerably to blood proteins. Likewise, FAVI does not bind to human serum albumin (HSA) and α1-acid glycoprotein (AGP) and shows relatively weak binding to the protein fraction of whole blood serum. Imatinib binds to AGP with high affinity (logK' = 5.8-6.0), while its binding to HSA is much weaker (logK' ≤ 4.0). The computed constants were used to model the distribution of IMA in blood plasma under physiological and 'acute-phase' conditions as well.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Unión Proteica , Mesilato de Imatinib/farmacología , SARS-CoV-2/metabolismo , Proteínas Sanguíneas/metabolismo , Orosomucoide/metabolismo , Albúmina Sérica Humana/metabolismo , Plasma/metabolismo
5.
Sci Rep ; 12(1): 5758, 2022 04 06.
Artículo en Inglés | MEDLINE | ID: covidwho-1778630

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causal agent of the COVID-19 pandemic. More than 274 million individuals have suffered from COVID-19 and over five million people have died from this disease so far. Therefore, there is an urgent need for therapeutic drugs. Repurposing FDA approved drugs should be favored since evaluation of safety and efficacy of de-novo drug design are both costly and time consuming. We report that imatinib, an Abl tyrosine kinase inhibitor, robustly decreases SARS-CoV-2 infection and uncover a mechanism of action. We show that imatinib inhibits the infection of SARS-CoV-2 and its surrogate lentivector pseudotype. In latter, imatinib inhibited both routes of viral entry, endocytosis and membrane-fusion. We utilized a system to quantify in real-time cell-cell membrane fusion mediated by the SARS-CoV-2 surface protein, Spike, and its receptor, hACE2, to demonstrate that imatinib inhibits this process in an Abl1 and Abl2 independent manner. Furthermore, cellular thermal shift assay revealed a direct imatinib-Spike interaction that affects Spike susceptibility to trypsin digest. Collectively, our data suggest that imatinib inhibits Spike mediated viral entry by an off-target mechanism. These findings mark imatinib as a promising therapeutic drug in inhibiting the early steps of SARS-CoV-2 infection.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Humanos , Mesilato de Imatinib/farmacología , Pandemias , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus
6.
Cell Metab ; 34(3): 424-440.e7, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1676683

RESUMEN

Coronavirus disease 2019 (COVID-19) represents a systemic disease that may cause severe metabolic complications in multiple tissues including liver, kidney, and cardiovascular system. However, the underlying mechanisms and optimal treatment remain elusive. Our study shows that impairment of ACE2 pathway is a key factor linking virus infection to its secondary metabolic sequelae. By using structure-based high-throughput virtual screening and connectivity map database, followed with experimental validations, we identify imatinib, methazolamide, and harpagoside as direct enzymatic activators of ACE2. Imatinib and methazolamide remarkably improve metabolic perturbations in vivo in an ACE2-dependent manner under the insulin-resistant state and SARS-CoV-2-infected state. Moreover, viral entry is directly inhibited by these three compounds due to allosteric inhibition of ACE2 binding to spike protein on SARS-CoV-2. Taken together, our study shows that enzymatic activation of ACE2 via imatinib, methazolamide, or harpagoside may be a conceptually new strategy to treat metabolic sequelae of COVID-19.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Mesilato de Imatinib/uso terapéutico , Enfermedades Metabólicas/tratamiento farmacológico , Metazolamida/uso terapéutico , SARS-CoV-2/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/complicaciones , COVID-19/metabolismo , COVID-19/virología , Células Cultivadas , Chlorocebus aethiops , Regulación hacia Abajo/efectos de los fármacos , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Mesilato de Imatinib/farmacología , Masculino , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/virología , Metazolamida/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Ratones Transgénicos , SARS-CoV-2/fisiología , Células Vero , Internalización del Virus/efectos de los fármacos
7.
J Cell Biochem ; 123(2): 155-160, 2022 02.
Artículo en Inglés | MEDLINE | ID: covidwho-1473858

RESUMEN

Drug repurposing is an attractive option for identifying new treatment strategies, in particular in extraordinary situations of urgent need such as the current coronavirus disease 2019 (Covid-19) pandemic. Recently, the World Health Organization announced testing of three drugs as potential Covid-19 therapeutics that are known for their dampening effect on the immune system. Thus, the underlying concept of selecting these drugs is to temper the potentially life-threatening overshooting of the immune system reacting to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. This viewpoint discusses the possibility that the impact of these and other drugs on autophagy contributes to their therapeutic effect by hampering the SARS-CoV-2 life cycle.


Asunto(s)
Antivirales/farmacología , Artesunato/farmacología , Autofagia/efectos de los fármacos , Tratamiento Farmacológico de COVID-19 , Reposicionamiento de Medicamentos , Mesilato de Imatinib/farmacología , Infliximab/farmacología , Pandemias , SARS-CoV-2/efectos de los fármacos , Antidepresivos/farmacología , Antivirales/uso terapéutico , Artesunato/uso terapéutico , Cloroquina/farmacología , Desarrollo de Medicamentos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/fisiología , Retículo Endoplásmico/virología , Endosomas/efectos de los fármacos , Endosomas/virología , Humanos , Hidroxicloroquina/farmacología , Mesilato de Imatinib/uso terapéutico , Infliximab/uso terapéutico , Membranas Intracelulares/efectos de los fármacos , Membranas Intracelulares/fisiología , Membranas Intracelulares/virología , Ivermectina/farmacología , Macrólidos/farmacología , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Niclosamida/farmacología , Niclosamida/uso terapéutico , ARN Viral/metabolismo , SARS-CoV-2/fisiología , Replicación Viral
8.
Int J Biol Macromol ; 190: 636-648, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1401500

RESUMEN

SARS-CoV-2 nucleocapsid (N) protein undergoes RNA-induced phase separation (LLPS) and sequesters the host key stress granule (SG) proteins, Ras-GTPase-activating protein SH3-domain-binding protein 1 and 2 (G3BP1 and G3BP2) to inhibit SG formation. This will allow viral packaging and propagation in host cells. Based on a genomic-guided meta-analysis, here we identify upstream regulatory elements modulating the expression of G3BP1 and G3BP2 (collectively called G3BP1/2). Using this strategy, we have identified FOXA1, YY1, SYK, E2F-1, and TGFBR2 as activators and SIN3A, SRF, and AKT-1 as repressors of G3BP1/2 genes. Panels of the activators and repressors were then used to identify drugs that change their gene expression signatures. Two drugs, imatinib, and decitabine have been identified as putative modulators of G3BP1/2 genes and their regulators, suggesting their role as COVID-19 mitigation agents. Molecular docking analysis suggests that both drugs bind to G3BP1/2 with a much higher affinity than the SARS-CoV-2 N protein. This study reports imatinib and decitabine as candidate drugs against N protein and G3BP1/2 protein.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Tratamiento Farmacológico de COVID-19 , Proteínas de la Nucleocápside de Coronavirus/química , ADN Helicasas/química , Decitabina/química , Mesilato de Imatinib/química , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Proteínas de Unión a Poli-ADP-Ribosa/química , ARN Helicasas/química , Proteínas con Motivos de Reconocimiento de ARN/química , Proteínas de Unión al ARN/química , SARS-CoV-2/química , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/metabolismo , COVID-19/metabolismo , Proteínas de la Nucleocápside de Coronavirus/metabolismo , ADN Helicasas/antagonistas & inhibidores , ADN Helicasas/metabolismo , Decitabina/farmacología , Sistemas de Liberación de Medicamentos , Genómica , Mesilato de Imatinib/farmacología , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/antagonistas & inhibidores , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , ARN Helicasas/antagonistas & inhibidores , ARN Helicasas/metabolismo , Proteínas con Motivos de Reconocimiento de ARN/antagonistas & inhibidores , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/metabolismo , SARS-CoV-2/metabolismo
10.
Antiviral Res ; 193: 105137, 2021 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1306846

RESUMEN

Following the emergence of SARS-CoV-2, the search for an effective and rapidly available treatment was initiated worldwide based on repurposing of available drugs. Previous reports described the antiviral activity of certain tyrosine kinase inhibitors (TKIs) targeting the Abelson kinase 2 against pathogenic coronaviruses. Imatinib, one of them, has more than twenty years of safe utilization for the treatment of hematological malignancies. In this context, Imatinib was rapidly evaluated in clinical trials against Covid-19. Here, we present the pre-clinical evaluation of imatinib in multiple models. Our results indicated that imatinib and another TKI, the masitinib, exhibit an antiviral activity in VeroE6 cells. However, imatinib was inactive in a reconstructed bronchial human airway epithelium model. In vivo, imatinib therapy failed to impair SARS-CoV-2 replication in a golden Syrian hamster model despite high concentrations in plasma and in the lung. Overall, these results do not support the use of imatinib and similar TKIs as antivirals in the treatment of Covid-19.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Mesilato de Imatinib/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , COVID-19/epidemiología , COVID-19/virología , Línea Celular , Chlorocebus aethiops , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos , Inhibidores Enzimáticos/farmacología , Epitelio , Femenino , Humanos , Pulmón/patología , Masculino , Mesocricetus , Células Vero , Replicación Viral/efectos de los fármacos
11.
Int J Mol Sci ; 22(13)2021 Jun 28.
Artículo en Inglés | MEDLINE | ID: covidwho-1288900

RESUMEN

A group of clinically approved cancer therapeutic tyrosine kinase inhibitors was screened to test their effects on the expression of angiotensin-converting enzyme 2 (ACE2), the cell surface receptor for SARS-CoV-2. Here, we show that the receptor tyrosine kinase inhibitor imatinib (also known as STI571, Gleevec) can inhibit the expression of the endogenous ACE2 gene at both the transcript and protein levels. Treatment with imatinib resulted in inhibition of cell entry of the viral pseudoparticles (Vpps) in cell culture. In FVB mice orally fed imatinib, tissue expression of ACE2 was reduced, specifically in the lungs and renal tubules, but not in the parenchyma of other organs such as the heart and intestine. Our finding suggests that receptor tyrosine kinases play a role in COVID-19 infection and can be therapeutic targets with combined treatments of the best conventional care of COVID-19.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Mesilato de Imatinib/farmacología , SARS-CoV-2/fisiología , Internalización del Virus/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/genética , Animales , COVID-19/patología , COVID-19/virología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Femenino , Genes Reporteros , Humanos , Ratones , Regiones Promotoras Genéticas , SARS-CoV-2/aislamiento & purificación
12.
Phys Chem Chem Phys ; 23(22): 12549-12558, 2021 Jun 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1233727

RESUMEN

The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) enters the host cell after the receptor binding domain (RBD) of the virus spike (S) glycoprotein binds to the human angiotensin-converting enzyme 2 (hACE2). This binding requires the RBD to undergo a conformational change from a closed to an open state. In the present study, a key pair of salt bridges formed by the side chains of K537 and E619, residues at the interfaces of SD1 and SD2, respectively, was identified to promote the opening of the RBD. Mutations of K537Q and E619D reduced their side chain lengths and eliminated this pair of salt bridges; as a result, the opening of the RBD was not observed in the MD simulations. Thus, blocking the formation of this pair of salt bridges is a promising approach for treating novel coronavirus disease 2019 (COVID-19). FDA approved drug molecules were screened by their capabilities of blocking the formation of the key pair of salt bridges, achieved by their positional stabilities in the cavity containing the side chains of K537 and E619 formed in the interface between SD1 and SD2. Simeprevir, imatinib, and naldemedine were identified to possess the desired capability with the most favorable interaction energies.


Asunto(s)
Antivirales/farmacología , Diseño de Fármacos , SARS-CoV-2/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Antivirales/química , Evaluación Preclínica de Medicamentos , Humanos , Mesilato de Imatinib/química , Mesilato de Imatinib/farmacología , Simulación del Acoplamiento Molecular , Naltrexona/análogos & derivados , Naltrexona/química , Naltrexona/farmacología , Dominios Proteicos/efectos de los fármacos , SARS-CoV-2/química , Simeprevir/química , Simeprevir/farmacología , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo
13.
Basic Clin Pharmacol Toxicol ; 128(4): 621-624, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: covidwho-965811

RESUMEN

Since the emergence of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) at the end of 2019, no vaccine has been approved to counter this infection and the available treatments are mainly directed against the immune pathology caused by the infection. The coronavirus disease 2019 (COVID-19) is currently causing a worldwide pandemic, pointing the urgent need for effective treatment. In such emergency, drug repurposing presents the best option for a rapid antiviral response. We assess here the in vitro activity of nilotinib, imatinib and dasatinib, three Abl tyrosine kinase inhibitors, against SARS-CoV-2. Although the last two compounds do not show antiviral efficacy, we observe inhibition with nilotinib in Vero-E6 cells and Calu-3 cells with EC50s of 1.44 µM and 3.06 µM, respectively. These values are close to the mean peak concentration of nilotinib observed at steady state in serum, making this compound a potential candidate for treatment of COVID-19 in vivo.


Asunto(s)
Antivirales/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , Línea Celular , Chlorocebus aethiops , Dasatinib/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Mesilato de Imatinib/farmacología , Técnicas In Vitro , Células Vero/virología
14.
Cell Death Dis ; 11(8): 656, 2020 08 19.
Artículo en Inglés | MEDLINE | ID: covidwho-725491

RESUMEN

The current epidemic of coronavirus disease-19 (COVID-19) caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) calls for the development of inhibitors of viral replication. Here, we performed a bioinformatic analysis of published and purported SARS-CoV-2 antivirals including imatinib mesylate that we found to suppress SARS-CoV-2 replication on Vero E6 cells and that, according to the published literature on other coronaviruses is likely to act on-target, as a tyrosine kinase inhibitor. We identified a cluster of SARS-CoV-2 antivirals with characteristics of lysosomotropic agents, meaning that they are lipophilic weak bases capable of penetrating into cells. These agents include cepharentine, chloroquine, chlorpromazine, clemastine, cloperastine, emetine, hydroxychloroquine, haloperidol, ML240, PB28, ponatinib, siramesine, and zotatifin (eFT226) all of which are likely to inhibit SARS-CoV-2 replication by non-specific (off-target) effects, meaning that they probably do not act on their 'official' pharmacological targets, but rather interfere with viral replication through non-specific effects on acidophilic organelles including autophagosomes, endosomes, and lysosomes. Imatinib mesylate did not fall into this cluster. In conclusion, we propose a tentative classification of SARS-CoV-2 antivirals into specific (on-target) versus non-specific (off-target) agents based on their physicochemical characteristics.


Asunto(s)
Betacoronavirus/fisiología , Infecciones por Coronavirus/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Neumonía Viral/metabolismo , Replicación Viral/efectos de los fármacos , Animales , Antivirales/farmacología , COVID-19 , Muerte Celular/efectos de los fármacos , Chlorocebus aethiops , Infecciones por Coronavirus/virología , Hidroxicloroquina/farmacología , Mesilato de Imatinib/farmacología , Lisosomas/efectos de los fármacos , Pandemias , Neumonía Viral/virología , Inhibidores de Proteínas Quinasas/farmacología , ARN Viral/efectos de los fármacos , SARS-CoV-2 , Células Vero , Carga Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA